2011
DOI: 10.1152/ajpheart.00703.2010
|View full text |Cite
|
Sign up to set email alerts
|

Endogenous HMGB1 contributes to ischemia-reperfusion-induced myocardial apoptosis by potentiating the effect of TNF-α/JNK

Abstract: High-mobility group box 1 (HMGB1) is a nuclear protein that has been implicated in the myocardial inflammation and injury induced by ischemia-reperfusion (I/R). The purpose of the present study was to assess the role of HMGB1 in myocardial apoptosis induced by I/R. In vivo, myocardial I/R induced an increase in myocardial HMGB1 expression and apoptosis. Inhibition of HMGB1 (A-box) ameliorated the I/R-induced myocardial apoptosis. In vitro, isolated cardiac myocytes were challenged with anoxia-reoxygenation (A/… Show more

Help me understand this report

Search citation statements

Order By: Relevance

Paper Sections

Select...
3
1

Citation Types

2
70
0
1

Year Published

2012
2012
2021
2021

Publication Types

Select...
8
1

Relationship

1
8

Authors

Journals

citations
Cited by 89 publications
(73 citation statements)
references
References 38 publications
2
70
0
1
Order By: Relevance
“…Hence, it is possible that upon release, HMGB1 directly or indirectly activates phospho-JNK pathway, which is supported by our results obtained due to the administration of rHMGB1 in this study. Moreover, recent studies have demonstrated that HMGB1 triggered a substantial potentiation of TNF-α-induced JNK phosphorylation and that inhibition of JNK (SP600125) prevented the myocyte apoptosis induced by a TNF-α/HMGB1 cocktail [29] ; these findings support our hypotheses.…”
Section: Discussionsupporting
confidence: 89%
See 1 more Smart Citation
“…Hence, it is possible that upon release, HMGB1 directly or indirectly activates phospho-JNK pathway, which is supported by our results obtained due to the administration of rHMGB1 in this study. Moreover, recent studies have demonstrated that HMGB1 triggered a substantial potentiation of TNF-α-induced JNK phosphorylation and that inhibition of JNK (SP600125) prevented the myocyte apoptosis induced by a TNF-α/HMGB1 cocktail [29] ; these findings support our hypotheses.…”
Section: Discussionsupporting
confidence: 89%
“…Because the anti-apoptotic effect of GL has been linked to the inhibition of HMGB1 and caspase-dependent cytochrome c release in vivo and ex vivo, the question that remains now concerns how GL regulates the BAX translocation and caspase-dependent cytochrome c release via the inhibition of HMGB1. The exact answer is currently unknown, but we speculate that GL may modulate the activity of a particular kinase, which contributes to BAX translocation and is involved in I/R-induced HMGB1-dependent apoptosis based on the following evidence: (1) Glycyrrhizin can alleviate HMGB1-induced hepatocyte apoptosis by inhibiting the p38-dependent mitochondrial pathway in Huh-BAT cells [27] ; (2) the JNK signaling pathway mediates Bax translocation and subsequent neuronal apoptosis through interaction with Bim after transient focal cerebral ischemia [20] ; (3) myocyte-derived HMGB1 and TNF-α work together to promote I/R-induced myocardial apoptosis through JNK activation [29] ; (4) blockade of HMGB1 using HMGB1 box A, another HMGB1 inhibitor, effectively decreased the phosphorylation of ERK1/2 and JNK, and finally reduced I/R-induced apoptosis injury [16] . The mitochondria-dependent apoptosis pathway is tightly regulated by the MAP kinase family, and JNK, ERK1/2, and p38 members of this family have been demonstrated to be activated in I/R injury [12,13] .…”
Section: Discussionmentioning
confidence: 99%
“…There exist growing recognition and experimental evidence to support that HMGB1 plays a pivotal role not only in the diseases such as sepsis, autoimmune disease, acute hepatic necrosis, acute lung injury 25, 26, 27, 28 but also in various heart diseases, including myocardial infarction and ischaemia‐reperfusion injury, however, with no consensus on the function of HMGB1 on the pathogenesis of the diseases 9, 16, 29, 30, 31, 32, 33, 34.…”
Section: Discussionmentioning
confidence: 99%
“…Internalized TNF-R2 via ASK1 can form a complex with AIP1, leading to JNK activation (15). Activated JNK signaling controls the release of cytochrome c and induces the apoptotic (38) and cardiac hypertrophic (36) programs. In our study, we found that CTSB deficiency blocked the pressure overload induced the release of TNF-␣ and activation of ASK1, JNK, and c-Jun (the proximal downstream product of JNK) and enhanced the release of cytochrome c. The JNK inhibitor SP600125 eliminated the activated JNK signaling provoked by TNF-␣ in CTSB overexpressed cardiomyocytes in response to ANG II.…”
mentioning
confidence: 99%